Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32441445

RESUMEN

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Asunto(s)
Antígenos CD40/metabolismo , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/virología , Virus ARN/fisiología , Transducción de Señal , Virosis/inmunología , Replicación Viral/fisiología , Enfermedad Aguda , Animales , Ligando de CD40/metabolismo , Ebolavirus/fisiología , Glicoproteínas/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-12/biosíntesis , Ratones Endogámicos C57BL , Modelos Biológicos , Peritoneo/patología , Peritoneo/virología , Factor 6 Asociado a Receptor de TNF/metabolismo , Virosis/virología
2.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30602604

RESUMEN

Gammaherpesviruses are ubiquitous viruses that establish lifelong infections. Importantly, these viruses are associated with numerous cancers and lymphoproliferative diseases. While risk factors for developing gammaherpesvirus-driven cancers are poorly understood, it is clear that elevated viral reactivation from latency often precedes oncogenesis. Here, we demonstrate that the liver X receptor alpha isoform (LXRα) restricts gammaherpesvirus reactivation in an anatomic-site-specific manner. We have previously demonstrated that deficiency of both LXR isoforms (α and ß) leads to an increase in fatty acid and cholesterol synthesis in primary macrophage cultures, with a corresponding increase in gammaherpesvirus replication. Interestingly, expression of fatty acid synthesis genes was not derepressed in LXRα-deficient hosts, indicating that the antiviral effects of LXRα are independent of lipogenesis. Additionally, the critical host defenses against gammaherpesvirus reactivation, virus-specific CD8+ T cells and interferon (IFN) signaling, remained intact in the absence of LXRα. Remarkably, using a murine gammaherpesvirus 68 (MHV68) reporter virus, we discovered that LXRα expression dictates the cellular tropism of MHV68 in the peritoneal cavity. Specifically, LXRα-/- mice exhibit reduced latency within the peritoneal B cell compartment and elevated latency within F4/80+ cells. Thus, LXRα restricts gammaherpesvirus reactivation through a novel mechanism that is independent of the known CD8+ T cell-based antiviral responses or changes in lipid synthesis and likely involves changes in the tropism of MHV68 in the peritoneal cavity.IMPORTANCE Liver X receptors (LXRs) are nuclear receptors that mediate cholesterol and fatty acid homeostasis. Importantly, as ligand-activated transcription factors, LXRs represent potential targets for the treatment of hypercholesterolemia and atherosclerosis. Here, we demonstrate that LXRα, one of the two LXR isoforms, restricts reactivation of latent gammaherpesvirus from peritoneal cells. As gammaherpesviruses are ubiquitous oncogenic agents, LXRs may represent a targetable host factor for the treatment of poorly controlled gammaherpesvirus infection and associated lymphomagenesis.


Asunto(s)
Linfocitos B/virología , Gammaherpesvirinae/inmunología , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/inmunología , Receptores X del Hígado/inmunología , Cavidad Peritoneal/virología , Latencia del Virus/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Femenino , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno/inmunología , Interferones/inmunología , Lipogénesis/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Peritoneo/inmunología , Peritoneo/virología , Transducción de Señal/inmunología , Tropismo/inmunología , Activación Viral/inmunología , Replicación Viral/inmunología
3.
Transpl Infect Dis ; 19(4)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28423469

RESUMEN

Among organ transplant recipients, cytomegalovirus (CMV) commonly results in various types of infection such as pneumonitis, hepatitis, and enterocolitis. However, CMV peritonitis is very rare and difficult to diagnose owing to lack of visible clinical signs. We present a case of a 35-year-old female kidney recipient who developed abdominal pain and urinary retention caused by CMV peritonitis. To our knowledge, this is the first case report of CMV peritonitis after organ transplantation to be diagnosed through histopathological examination.


Asunto(s)
Infecciones por Citomegalovirus/patología , Citomegalovirus/aislamiento & purificación , Trasplante de Riñón/efectos adversos , Peritoneo/virología , Peritonitis/patología , Adulto , Infecciones por Citomegalovirus/diagnóstico por imagen , Infecciones por Citomegalovirus/etiología , Infecciones por Citomegalovirus/virología , Femenino , Humanos , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía , Peritoneo/patología , Peritonitis/diagnóstico por imagen , Peritonitis/etiología , Peritonitis/virología , Teratoma/patología , Teratoma/cirugía , Tomografía Computarizada por Rayos X
4.
Fish Shellfish Immunol ; 64: 210-217, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28302579

RESUMEN

IgM+ B cells have been recently demonstrated to be key regulators of peritoneal inflammation in teleost, as a large number of them occupy the peritoneal cavity after 48 h of antigenic stimulation. Despite this, the number of studies addressing the mechanism through which this cell population expands and differentiates in response to stimuli has been scarcely addressed. Because the BAFF/APRIL axis is known to play a major role in B cell survival and differentiation in mammals, we hypothesized that it could be affected in the peritoneal cavity in response to an inflammatory stimulus. To verify this hypothesis, we studied how BAFF, APRIL and the fish-specific related cytokine BALM as well as their putative receptors are regulated in rainbow trout after intraperitoneal (i.p.) injection of viral hemorrhagic septicemia virus (VHSV). When the transcriptional analysis was performed in total cells from the peritoneum, we observed that VHSV provoked an up-regulation of both BAFF and BAFF receptor (BAFF-R) mRNA levels. However, when we examined how isolated peritoneal IgM+ B cells were transcriptionally affected by VHSV i.p. injection, we found that APRIL, BALM and the transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) were also up-regulated in response to the virus. IgM- cells, on the other hand, only up-regulated BALM transcription in response to VHSV. Finally, to gain further insight on the role that these cytokines play in the peritoneum, we have studied their effect on the survival of peritoneal IgM+ B cells. This work demonstrates a key role for the BAFF/APRIL axis in the peritoneal inflammatory response and contributes to further understanding how IgM+ B cells are regulated at this specific peripheral site.


Asunto(s)
Enfermedades de los Peces/genética , Proteínas de Peces/genética , Novirhabdovirus/fisiología , Oncorhynchus mykiss , Infecciones por Rhabdoviridae/veterinaria , Animales , Receptor del Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/metabolismo , Linfocitos B/inmunología , Citocinas/genética , Citocinas/metabolismo , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Proteínas de Peces/metabolismo , Peritoneo/fisiopatología , Peritoneo/virología , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/virología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
5.
J Virol ; 90(20): 9330-7, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27512061

RESUMEN

UNLABELLED: Type I interferons (IFNs) crucially contribute to host survival upon viral infections. Robust expression of type I IFNs (IFN-α/ß) and induction of an antiviral state critically depend on amplification of the IFN signal via the type I IFN receptor (IFNAR). A small amount of type I IFN produced early upon virus infection binds the IFNAR and activates a self-enhancing positive feedback loop, resulting in induction of large, protective amounts of IFN-α. Unexpectedly, we found robust, systemic IFN-α expression upon infection of IFNAR knockout mice with the orthomyxovirus Thogoto virus (THOV). The IFNAR-independent IFN-α production required in vivo conditions and was not achieved during in vitro infection. Using replication-incompetent THOV-derived virus-like particles, we demonstrate that IFNAR-independent type I IFN induction depends on viral polymerase activity but is largely independent of viral replication. To discover the cell type responsible for this effect, we used type I IFN reporter mice and identified CD11b(+) F4/80(+) myeloid cells within the peritoneal cavity of infected animals as the main source of IFNAR-independent type I IFN, corresponding to the particular tropism of THOV for this cell type. IMPORTANCE: Type I IFNs are crucial for the survival of a host upon most viral infections, and, moreover, they shape subsequent adaptive immune responses. Production of protective amounts of type I IFN critically depends on the positive feedback amplification via the IFNAR. Unexpectedly, we observed robust IFNAR-independent type I IFN expression upon THOV infection and unraveled molecular mechanisms and determined the tissue and cell type involved. Our data indicate that the host can effectively use alternative pathways to induce type I IFN responses if the classical feedback amplification is not available. Understanding how type I IFN can be produced in large amounts independently of IFNAR-dependent enhancement will identify mechanisms which might contribute to novel therapeutic strategies to fight viral pathogens.


Asunto(s)
Antígeno CD11b/metabolismo , Interferón Tipo I/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Peritoneo/virología , Receptor de Interferón alfa y beta/metabolismo , Thogotovirus/metabolismo , Animales , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritoneo/metabolismo , Transducción de Señal/fisiología , Replicación Viral/fisiología
6.
Virology ; 492: 140-4, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26945150

RESUMEN

Gammaherpesviruses establish life-long infection in most adults and are associated with the development of B cell lymphomas. While the interaction between gammaherpesviruses and splenic B cells has been explored, very little is known about gammaherpesvirus infection of B-1 B cells, innate-like B cells that primarily reside in body cavities. This study demonstrates that B-1 B cells harbor the highest frequency of latently infected cells in the peritoneum throughout chronic infection, highlighting a previously unappreciated feature of gammaherpesvirus biology.


Asunto(s)
Subgrupos de Linfocitos B/virología , Gammaherpesvirinae/fisiología , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno , Células Mieloides/virología , Bazo/virología , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/inmunología , Células Mieloides/patología , Peritoneo/inmunología , Peritoneo/patología , Peritoneo/virología , Bazo/inmunología , Bazo/patología , Activación Viral , Latencia del Virus
7.
J Immunol ; 195(2): 576-86, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26062996

RESUMEN

Nonclassical MHC class Ib-restricted invariant T (iT) cell subsets are attracting interest because of their potential to regulate immune responses against various pathogens. The biological relevance and evolutionary conservation of iT cells have recently been strengthened by the identification of iT cells (invariant Vα6 [iVα6]) restricted by the nonclassical MHC class Ib molecule XNC10 in the amphibian Xenopus laevis. These iVα6 T cells are functionally similar to mammalian CD1d-restricted invariant NKT cells. Using the amphibian pathogen frog virus 3 (FV3) in combination with XNC10 tetramers and RNA interference loss of function by transgenesis, we show that XNC10-restricted iVα6 T cells are critical for early antiviral immunity in adult X. laevis. Within hours following i.p. FV3 infection, iVα6 T cells were specifically recruited from the spleen into the peritoneum. XNC10 deficiency and concomitant lack of iVα6 T cells resulted in less effective antiviral and macrophage antimicrobial responses, which led to impaired viral clearance, increased viral dissemination, and more pronounced FV3-induced kidney damage. Together, these findings imply that X. laevis XNC10-restricted iVα6 T cells play important roles in the early anti-FV3 response and that, as has been suggested for mammalian invariant NKT cells, they may serve as immune regulators polarizing macrophage effector functions toward more effective antiviral states.


Asunto(s)
Proteínas Anfibias/inmunología , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Innata , Ranavirus/inmunología , Linfocitos T/inmunología , Proteínas Anfibias/antagonistas & inhibidores , Proteínas Anfibias/genética , Animales , Movimiento Celular , Infecciones por Virus ADN/patología , Infecciones por Virus ADN/virología , Femenino , Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Inmunofenotipificación , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/patología , Células T Asesinas Naturales/virología , Peritoneo/inmunología , Peritoneo/patología , Peritoneo/virología , Multimerización de Proteína , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Transducción de Señal , Bazo/inmunología , Bazo/patología , Bazo/virología , Linfocitos T/patología , Linfocitos T/virología , Xenopus laevis
8.
J Virol ; 87(22): 12151-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23986598

RESUMEN

Promyelocytic leukemia protein (PML) is an essential organizer of PML nuclear bodies (NBs), which carry out a variety of activities, including antiviral functions. Herpesviruses from all subfamilies encode proteins that counteract PML NB-mediated antiviral defenses by multiple mechanisms. However, because of the species specificity of herpesviruses, only a limited number of in vivo studies have been undertaken to investigate the effect of PML or PML NBs on herpesvirus infection. To address this central issue in herpesvirus biology, we studied the course of infection in wild-type and PML⁻/⁻ mice using murine gammaherpesvirus 68 (MHV68), which encodes a tegument protein that induces PML degradation. While acute infection in PML⁻/⁻ mice progressed similarly to that in wild-type mice, the lytic reactivation frequency was higher in peritoneal exudate cells, due to both an increase of MHV68 genome-positive cells and greater reactivation efficiency. We also detected a higher frequency of persistent infection in PML⁻/⁻ peritoneal cells. These findings suggest that the PML protein can repress the establishment or maintenance of gammaherpesvirus latency in vivo. Further use of the PML⁻/⁻ mouse model should aid in dissecting the molecular mechanisms that underlie the role of PML in gammaherpesvirus latency and may yield clues for how PML modulates herpesvirus latency in general.


Asunto(s)
Embrión de Mamíferos/virología , Fibroblastos/virología , Infecciones por Herpesviridae/virología , Proteínas Nucleares/fisiología , Peritoneo/virología , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Latencia del Virus , Replicación Viral , Animales , Western Blotting , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Citometría de Flujo , Gammaherpesvirinae/patogenicidad , Regulación Viral de la Expresión Génica , Genoma Viral , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Ratones , Ratones Noqueados , Peritoneo/metabolismo , Peritoneo/patología , Proteína de la Leucemia Promielocítica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
PLoS One ; 6(6): e20169, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695116

RESUMEN

Several new immunodeficient mouse models for human cell engraftment have recently been introduced that include the Rag2(-/-)γc(-/-), NOD/SCID, NOD/SCIDγc(-/-) and NOD/SCIDß2m(-/-) strains. Transplantation of these mice with CD34(+) human hematopoietic stem cells leads to prolonged engraftment, multilineage hematopoiesis and the capacity to generate human immune responses against a variety of antigens. However, the various mouse strains used and different methods of engrafting human cells are beginning to illustrate strain specific variations in engraftment levels, duration and longevity of mouse life span. In these proof-of-concept studies we evaluated the Balb/c-Rag1(-/-)γ(-/-) strain for engraftment by human fetal liver derived CD34(+) hematopoietic cells using the same protocol found to be effective for Balb/c-Rag2(-/-)γc(-/-) mice. We demonstrate that these mice can be efficiently engrafted and show multilineage human hematopoiesis with human cells populating different lymphoid organs. Generation of human cells continues beyond a year and production of human immunoglobulins is noted. Infection with HIV-1 leads to chronic viremia with a resultant CD4 T cell loss. To mimic the predominant sexual viral transmission, we challenged humanized Rag1(-/-)γc(-/-) mice with HIV-1 via vaginal route which also resulted in chronic viremia and helper T cell loss. Thus these mice can be further exploited for studying human pathogens that infect the human hematopoietic system in an in vivo setting.


Asunto(s)
Linaje de la Célula , Infecciones por VIH/inmunología , VIH-1/inmunología , Hematopoyesis , Proteínas de Homeodominio/metabolismo , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Vagina/virología , Animales , Formación de Anticuerpos/inmunología , Antígenos CD34/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Susceptibilidad a Enfermedades/inmunología , Femenino , Citometría de Flujo , VIH-1/patogenicidad , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoglobulinas/metabolismo , Inmunohistoquímica , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Tejido Linfoide/citología , Ratones , Membrana Mucosa/virología , Peritoneo/virología
10.
J Gen Virol ; 90(Pt 6): 1461-1470, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264614

RESUMEN

Gammaherpesviruses infect at least 90 % of the world's population. Infection control is difficult, in part because some fundamental features of host colonization remain unknown, for example whether normal latency establishment requires viral lytic functions. Since human gammaherpesviruses have narrow species tropisms, answering such questions requires animal models. Murid herpesvirus-4 (MuHV-4) provides one of the most tractable. MuHV-4 genomes delivered to the lung or peritoneum persist without lytic replication. However, they fail to disseminate systemically, suggesting that the outcome is inoculation route-dependent. After upper respiratory tract inoculation, MuHV-4 infects mice without involving the lungs or peritoneum. We examined whether host entry by this less invasive route requires the viral thymidine kinase (TK), a gene classically essential for lytic replication in terminally differentiated cells. MuHV-4 TK knockouts delivered to the lung or peritoneum were attenuated but still reached lymphoid tissue. In contrast, TK knockouts delivered to the upper respiratory tract largely failed to establish a detectable infection. Therefore TK, and by implication lytic replication, is required for MuHV-4 to establish a significant infection by a non-invasive route.


Asunto(s)
Infecciones por Herpesviridae/virología , Rhadinovirus/crecimiento & desarrollo , Timidina Quinasa/fisiología , Infecciones Tumorales por Virus/virología , Proteínas Virales/fisiología , Animales , Femenino , Técnicas de Inactivación de Genes , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Peritoneo/virología , Sistema Respiratorio/virología , Timidina Quinasa/deficiencia
11.
J Mol Med (Berl) ; 87(4): 385-99, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19139835

RESUMEN

The dissemination of malignant gastric cells to the peritoneum occurs frequently, usually as an early event in disease, and results in poor patient prognosis. Surgery and chemotherapy offer limited therapeutic success. The low-pathogenic human enterovirus, Echovirus 1 (EV1), is an oncolytic virus that selectively targets and destroys malignant prostate and ovarian cancer xenografts in vivo. Lytic EV1 infection requires the cell surface expression of alpha(2)beta(1), an integrin involved in the dissemination of gastric cancer cells to the peritoneum. Herein, we evaluated the capacity of EV1 for anti-neoplastic cell action in gastric peritoneal carcinomatosis. Flow cytometric analysis demonstrated that alpha(2)beta(1) was abundantly surface expressed on a panel of gastric cancer cell lines, rendering the majority of lines highly susceptible to in vitro lytic EV1 infection and supportive of efficient viral progeny production. A bioluminescent MKN-45-Luc SCID mouse model of peritoneal dissemination was developed to allow real-time non-invasive monitoring of peritoneal tumor burden. Employing this mouse model, we demonstrated a therapeutic dose-response for escalating oncolytic EV1 doses. Taken together, these results emphasize the exciting potential for EV1 as a single or adjunct therapy for the control of the peritoneal dissemination of gastric cancer.


Asunto(s)
Enterovirus Humano B/fisiología , Neoplasias Peritoneales/terapia , Neoplasias Gástricas/terapia , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Integrina alfa2beta1/análisis , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/virología , Peritoneo/metabolismo , Peritoneo/patología , Peritoneo/virología , Neoplasias Gástricas/patología , Neoplasias Gástricas/virología , Análisis de Supervivencia , Transfección , Carga Tumoral
12.
Clin Exp Immunol ; 155(1): 28-34, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19076826

RESUMEN

The capacity of the immunomodulatory drug rapamycin (RAPA) to inhibit replication of the CCR5 strain of human immunodeficiency virus (HIV) in vitro prompted us to test its effects in a murine preclinical model of HIV infection. RAPA (0.6 or 6 mg/kg body weight) or its vehicle were administered daily, per os, to SCID mice reconstituted with human peripheral blood leucocytes (hu-PBL) starting 2 days before the intraperitoneal challenge with the R5 tropic SF162 strain of HIV-1 (1000 50% tissue culture infective dose/ml). Relative to hu-PBL-SCID mice that received no treatment, HIV-infected hu-PBL-SCID mice treated with the vehicle control for 3 weeks exhibited a severe depletion of CD4(+) cells (90%), an increase in CD8(+) cells and an inversion of the CD4(+)/CD8(+) cell ratio. In contrast, treatment of HIV-infected mice with RAPA prevented a decrease in CD4(+) cells and the increase of CD8(+) cells, thereby preserving the original CD4(+):CD8(+) cell ratio. Viral infection also resulted in the detection of HIV-DNA within peritoneal cells and spleen, and lymph node tissues of the vehicle-treated mice within 3 weeks of the viral challenge. In contrast, treatment with RAPA decreased cellular provirus integration and reduced HIV-RNA levels in the blood. Furthermore, in co-cultivation assays, spleens from RAPA-treated mice exhibited a reduced capacity for infecting allogeneic T cells which was dose-dependent. These data show that RAPA possesses powerful anti-viral activity against R5 strains of HIV in vivo and support the use of additional studies to evaluate the potential application of this drug in the management of HIV patients.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , VIH-1 , Inmunosupresores/uso terapéutico , Sirolimus/uso terapéutico , Animales , Relación CD4-CD8 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cocultivo , ADN Viral/análisis , Humanos , Ganglios Linfáticos/virología , Ratones , Ratones SCID , Modelos Animales , Peritoneo/virología , ARN Viral/sangre , Bazo/virología
13.
Virol J ; 5: 25, 2008 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-18267012

RESUMEN

BACKGROUND: La Crosse virus (LACV), family Bunyaviridae, was first identified as a human pathogen in 1960 after its isolation from a 4 year-old girl with fatal encephalitis in La Crosse, Wisconsin. LACV is a major cause of pediatric encephalitis in North America and infects up to 300,000 persons each year of which 70-130 result in severe disease of the central nervous system (CNS). As an initial step in the establishment of useful animal models to support vaccine development, we examined LACV infectivity, pathogenesis, and immunogenicity in both weanling mice and rhesus monkeys. RESULTS: Following intraperitoneal inoculation of mice, LACV replicated in various organs before reaching the CNS where it replicates to high titer causing death from neurological disease. The peripheral site where LACV replicates to highest titer is the nasal turbinates, and, presumably, LACV can enter the CNS via the olfactory neurons from nasal olfactory epithelium. The mouse infectious dose50 and lethal dose50 was similar for LACV administered either intranasally or intraperitoneally. LACV was highly infectious for rhesus monkeys and infected 100% of the animals at 10 PFU. However, the infection was asymptomatic, and the monkeys developed a strong neutralizing antibody response. CONCLUSION: In mice, LACV likely gains access to the CNS via the blood stream or via olfactory neurons. The ability to efficiently infect mice intranasally raises the possibility that LACV might use this route to infect its natural hosts. Rhesus monkeys are susceptible to LACV infection and develop strong neutralizing antibody responses after inoculation with as little as 10 PFU. Mice and rhesus monkeys are useful animal models for LACV vaccine immunologic testing although the rhesus monkey model is not optimal.


Asunto(s)
Modelos Animales de Enfermedad , Encefalitis de California/virología , Virus La Crosse/inmunología , Virus La Crosse/patogenicidad , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Línea Celular , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Chlorocebus aethiops , Femenino , Humanos , Virus La Crosse/fisiología , Dosificación Letal Mediana , Macaca mulatta , Ratones , Pruebas de Neutralización , Nariz/virología , Peritoneo/virología , Células Vero , Replicación Viral
14.
J Immunol ; 179(12): 8392-402, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056385

RESUMEN

The human gamma-herpesviruses, EBV and Kaposi's sarcoma-associated herpesvirus, are widely disseminated and are associated with the onset of a variety of malignancies. Thus, the development of prophylactic and therapeutic vaccination strategies is an important goal. The experimental mouse gamma-herpesvirus, gammaHV68 (or MHV-68), has provided an in vivo model for studying immune control of these persistent viruses. In the current studies, we have examined infectivity, immunogenicity, and protective efficacy following infection with a replication-deficient gammaHV68 blocked in late viral gene expression, ORF31STOP. The data show that ORF31STOP was able to latently infect B cells. However, the anatomical site and persistence of the infection depended on the route of inoculation, implicating a role for viral replication in viral spread but not the infectivity per se. Furthermore, i.p. infection with ORF31STOP elicited strong cellular immunity but a non-neutralizing Ab response. In contrast, intranasal infection was poorly immunogenic. Consistent with this, mice infected i.p. had enhanced control of both the lytic and latent viral loads following challenge with wild-type gammaHV68, whereas intranasal infected mice were not protected. These data provide important insight into mechanisms of infection and protective immunity for the gamma-herpesviruses and demonstrate the utility of replication-deficient mutant viruses in direct testing of "proof of principal" vaccination strategies.


Asunto(s)
Gammaherpesvirinae/genética , Gammaherpesvirinae/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Vacunas contra Herpesvirus/genética , Vacunas contra Herpesvirus/inmunología , Animales , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Linfocitos B/virología , Gammaherpesvirinae/fisiología , Regulación Viral de la Expresión Génica , Vacunas contra Herpesvirus/uso terapéutico , Inmunidad Celular , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peritoneo/inmunología , Peritoneo/virología , Bazo/inmunología , Bazo/virología , Carga Viral , Latencia del Virus/genética , Replicación Viral/genética
15.
Vopr Virusol ; 49(2): 11-7, 2004.
Artículo en Ruso | MEDLINE | ID: mdl-15106377

RESUMEN

Reproduction of the Ebola strains (ES) virus causing lethality in guinea pigs as well as in peritoneal macrophages and aorta explants of animals was investigated in vitro and in vivo; besides, production of interferon-gamma (IFN-gamma) and of tumor necrosis factor-alpha (TNF-alpha) by macrophages and endotheliocytes of guinea pigs was also studied. The interplay "macrophage--ES" by the example of 2 models of susceptibility to ES demonstrates that the ES lethality is not unambiguously related only with a level of virus reproduction in macrophages. The interplay "endotheliocyte--ES" is indicative of that the ES lethality is inversely dependent on a level of production of the IFN-gamma and of TNF-alpha by endotheliocytes. In general, the Eboly fever lethality is not conditioned only by the ability or inability of ES to reproduce in macrophages and endotheliocytes; it also depends on a variety of pathogenetic factors, one of which could be the cytotoxic action of immune complexes shaping in the process of infection progression.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/virología , Replicación Viral , Adaptación Fisiológica , Animales , Aorta/inmunología , Aorta/virología , Recuento de Células , Técnicas de Cultivo , Células Endoteliales/inmunología , Células Endoteliales/virología , Cobayas , Fiebre Hemorrágica Ebola/inmunología , Interferón gamma/análisis , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/virología , Peritoneo/inmunología , Peritoneo/virología , Especificidad de la Especie , Factor de Necrosis Tumoral alfa/análisis
16.
Nephrol Dial Transplant ; 19(5): 1293-7, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14993505

RESUMEN

BACKGROUND: Initial studies of adenovirus-mediated gene transfer to the peritoneum have shown transgene expression in the mesothelium from the parietal peritoneum. Using a replication-deficient adenovirus encoding beta-galactosidase (Ad beta Gal), we investigated the expression efficiency and the distribution of the transgene to different areas of both visceral and parietal peritoneum and to extra-peritoneal tissues. METHODS: Male Wistar rats received an intraperitoneal injection of 15 ml of 0.9% NaCl alone or containing 1 x 10(9) or 3 x 10(9) p.f.u. of Ad beta Gal. Evaluations of the histology of the peritoneum, the transgene expression and the safety of adenovirus-mediated gene transfer, using measurement of both beta Gal activity and staining, were performed 1, 3 and 5 days post-injection. RESULTS: At 1 day post-injection of 3 x 10(9) p.f.u. of Ad beta Gal, significant beta Gal activity and staining were detected in the omentum and mesenteric peritoneum. beta Gal staining was observed in endothelial cells, mesothelial cells and adipocytes. Focal mononuclear infiltrates restricted to the submesothelial area of the visceral peritoneum were also observed. No expression was detected in the mesocolon and parietal peritoneum, where the mesothelium was damaged. Significant beta Gal activity and staining were observed in lymph nodes, lungs, liver, heart and kidneys, in the absence of inflammatory changes. CONCLUSIONS: Intraperitoneal delivery of adenoviral vectors allows highly efficient transgene expression in mesothelial cells, but also in endothelial cells and adipocytes of the visceral peritoneum. Adverse focal mononuclear infiltrates, as well as spreading of the adenoviral vector from the abdominal cavity to the systemic circulation, were observed in parallel. Transgene expression in endothelial cells is potentially important since the latter play a key role in the alterations of the peritoneal membrane associated with long-term peritoneal dialysis. However, these data emphasize the need for less immunogenic adenoviral vectors, ideally containing an endothelial cell-specific promoter, to overcome immune response-related problems and spreading to extra-peritoneal tissues.


Asunto(s)
Adenoviridae/genética , Peritoneo/virología , Adipocitos/virología , Animales , Células Endoteliales/virología , Epitelio/virología , Técnicas de Transferencia de Gen , Vectores Genéticos , Masculino , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Recombinación Genética , Transfección , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
17.
J Immunol ; 172(5): 3139-50, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14978120

RESUMEN

Viral infections have been shown to induce lymphopenias that lower memory CD8 T cell frequencies, and they also have been shown to cause a permanent loss of memory cells specific to previously encountered pathogens. In this study, the patterns and significance of virus-induced memory CD8 T cell depletion were examined in mice immune to heterologous (Pichinde, vesicular stomatitis, vaccinia) viruses and subsequently challenged with acute or persistent lymphocytic choriomeningitis virus infections. Memory CD8 T cell loss was comprehensive and occurred in both lymphoid and peripheral tissues of the immune host. The impact of the loss of memory T cells was reflected by in vivo cytotoxicity assays, which showed decreased clearance of epitope-expressing targets. Memory CD8 T cell loss occurred very early (day 2) after infection, and was thereafter sustained, consistent more with an active deletion model than with a competition model. Cross-reactive T cells, in contrast, increased in number, but memory cells were reduced whether or not there was competition from cross-reactive T cells. Memory T cell loss was more profound during persistent infection than after acute infection. Adoptive transfer studies showed that, unlike the resolved acute infection, in which the reduced memory frequencies became stable, memory T cell loss was a continuously ongoing process during persistent infection. This study therefore links an early virus-induced lymphopenia to a subsequent long-term loss of CD8 T cell memory and offers a new mechanism for immune deficiency during persistent viral infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Linfopenia/inmunología , Linfopenia/virología , Subgrupos de Linfocitos T/inmunología , Enfermedad Aguda , Animales , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Supervivencia Celular/inmunología , Enfermedad Crónica , Citotoxicidad Inmunológica , Epítopos de Linfocito T/inmunología , Cinética , Recuento de Linfocitos , Coriomeningitis Linfocítica/patología , Linfopenia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/química , Proteínas Nucleares/inmunología , Peritoneo/inmunología , Peritoneo/virología , Virus Pichinde/inmunología , Subgrupos de Linfocitos T/patología , Subgrupos de Linfocitos T/virología , Factores de Tiempo , Virus Vaccinia/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Proteínas Virales/química , Proteínas Virales/inmunología
18.
J Virol ; 76(3): 1154-62, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11773391

RESUMEN

Pichinde virus is an arenavirus that infects guinea pigs and serves as an animal model for human Lassa fever. An attenuated Pichinde virus variant (P2) and a virulent variant (P18) are being used to delineate pathogenic mechanisms that culminate in shock. In guinea pigs, the infection has been shown to begin in peritoneal macrophages following intraperitoneal inoculation and then spreads to the spleen and other reticuloendothelial organs. We show here that infection of the murine monocytic cell line P388D1 with either Pichinde virus variant resulted in the induction of inflammatory cytokines and effectors, including interleukin-6 and tumor necrosis factor alpha. Since these genes are regulated in part by the cellular transcription factors NF-kappaB and RBP-Jkappa, we compared the activities of NF-kappaB and RBP-Jkappa in P388D1 cells following infection with Pichinde virus. The attenuated P2 virus inhibited NF-kappaB activation and caused a shift in the size of the RBP-Jkappa complex. The virulent P18 virus showed less inhibition of NF-kappaB and failed to alter the size of the RBP-Jkappa complex. Peritoneal cells from P2-infected guinea pigs showed induction of NF-kappaB RelA/p50 heterodimer and p50/p50 homodimer and manifested an increase in the size of RBP-Jkappa. By contrast, P18 induced large amounts of the NF-kappaB p50/p50 dimer but failed to induce RelA/p50 or to cause an increase in the RBP-Jkappa size. Taken together, these changes suggest that the attenuated viral strain induces an "activation" of macrophages, while the virulent form of the virus does not.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Proteínas de Unión al ADN/biosíntesis , Macrófagos Peritoneales/inmunología , Macrófagos/inmunología , FN-kappa B/biosíntesis , Proteínas Nucleares , Virus Pichinde/inmunología , Animales , Infecciones por Arenaviridae/virología , Línea Celular , Núcleo Celular/metabolismo , Cobayas , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas , Interleucina-6/biosíntesis , Macrófagos/virología , Macrófagos Peritoneales/virología , Masculino , Ratones , FN-kappa B/genética , Peritoneo/citología , Peritoneo/virología , Virus Pichinde/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Proc Natl Acad Sci U S A ; 97(6): 2725-30, 2000 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-10694575

RESUMEN

Mice that lack CD4(+) T cells remain clinically normal for more than 60 days after respiratory challenge with the murine gamma-herpesvirus 68 (gammaHV-68), then develop symptoms of a progressive wasting disease. The gammaHV-68-specific CD8(+) T cells that persist in these I-A(b-/-) mice are unable to prevent continued, but relatively low level, virus replication. Postexposure challenge with recombinant vaccinia viruses expressing gammaHV-68 lytic cycle epitopes massively increased the magnitude of the gammaHV-68-specific CD8(+) population detectable by staining with tetrameric complexes of MHC class I glycoprotein + peptide, or by interferon-gamma production subsequent to in vitro restimulation with peptide. The boosting effect was comparable for gammaHV-68-infected I-A(b-/-) and I-A(b+/+) mice within 7 days of challenge, and took more than 110 days to return to prevaccination levels in the I-A(b+/+) controls. Although the life-span of the I-A(b-/-) mice was significantly increased, there was no effect on long-term survival. A further boost with a recombinant influenza A virus failed to improve the situation. Onset of weight loss was associated with a decline in gammaHV-68-specific CD8(+) T cell numbers, though it is not clear whether this was a cause or an effect of the underlying pathology. Even very high levels of virus-specific CD8(+) T cells thus provide only transient protection against the uniformly lethal consequences of gammaHV-68 infection under conditions of CD4(+) T cell deficiency.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Gammaherpesvirinae/inmunología , Vacunas Virales/uso terapéutico , Animales , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Femenino , Citometría de Flujo , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Peritoneo/inmunología , Peritoneo/virología , Bazo/inmunología , Bazo/virología , Factores de Tiempo , Virus Vaccinia/metabolismo
20.
Proc Natl Acad Sci U S A ; 96(19): 10881-6, 1999 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-10485920

RESUMEN

Chemokine homologs are encoded by many large DNA viruses, suggesting that they contribute to control of host leukocyte transmigration and trafficking during viral infection. Murine cytomegalovirus carries a CC (beta) chemokine homolog gene giving rise to two related proteins, murine cytomegalovirus chemokine 1 and 2 (MCK-1 and MCK-2). MCK-1 peptide was found to induce calcium signaling and adherence in murine peritoneal macrophages. Cells bearing human chemokine receptor CCR3 and the human macrophage THP1 cell line were responsive to MCK-1. This pattern suggested that MCK-1 might act as an agonist, promoting leukocyte trafficking during viral infection. Consistent with this prediction, MCK-1/MCK-2 mutant viruses exhibit dramatically reduced peak levels of monocyte-associated viremia in experimentally infected mice. Thus, MCK-1/MCK-2 appears to promote host leukocyte migration to initial sites of infection and may be responsible for attracting monocytes or macrophages that efficiently disseminate virus in the host.


Asunto(s)
Quimiocinas CC/fisiología , Quimiocinas/genética , Quimiocinas/metabolismo , Citomegalovirus/genética , Monocitos/metabolismo , Proteínas Virales , Viremia/genética , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Quimiocinas CC/genética , Citomegalovirus/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Modelos Genéticos , Datos de Secuencia Molecular , Monocitos/virología , Peritoneo/metabolismo , Peritoneo/virología , Recombinación Genética , Homología de Secuencia de Aminoácido , Factores de Tiempo , Viremia/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...